Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Acta Biomater ; 181: 222-234, 2024 06.
Article in English | MEDLINE | ID: mdl-38648912

ABSTRACT

Polymeric biomedical implants are an important clinical tool, but degradation remains difficult to determine post-implantation. Computed tomography (CT) could be a powerful tool for device monitoring, but polymers require incorporation of radiopaque contrast agents to be distinguishable from tissue. In addition, immune response to radiopaque devices must be characterized as it modulates device function. Radiopaque devices and films were produced by incorporating 0-20 wt% TaOx nanoparticles into polymers: polycaprolactone (PCL) and poly(lactide-co-glycolide) (PLGA). In vitro inflammatory responses of mouse bone marrow-derived macrophages to polymer matrix incorporating TaOx nanoparticles was determined by monitoring cytokine secretion. Nanoparticle addition stimulated a slight inflammatory reaction, increasing TNFα secretion, mediated by changes in polymer matrix properties. Subsequently, devices (PLGA 50:50 + 20 wt% TaOx) were implanted subcutaneously in a mouse model of chronic inflammation, that featured a sustained increase in inflammatory response local to the implant site over 12 weeks. No changes to device degradation rates or foreign body response were noted between a normal and chronically stimulated inflammatory environment. Serial CT device monitoring post-implantation provided a detailed timeline of device collapse, with no rapid, spontaneous release of nanoparticles that occluded matrix visualization. Importantly, repeat CT sessions did not ablate the immune system or alter degradation kinetics. Thus, polymer devices incorporating radiopaque nanoparticles can be used for in situ monitoring and be readily combined with other medical imaging techniques, for a dynamic view biomaterial and tissue interactions. STATEMENT OF SIGNIFICANCE: A growing number of implantable devices are in use in the clinic, exposing patients to inherent risks of implant movement, collapse, and infection. The ability to monitor implanted devices would enable faster diagnosis of failure and open the door for personalized rehabilitation therapies - both of which could vastly improve patient outcomes. Unfortunately, polymeric materials which make up most biomedical devices are not radiologically distinguishable from tissue post-implantation. The introduction of radiopaque nanoparticles into polymers allows for serial monitoring via computed tomography, without affecting device degradation. Here we demonstrate for the first time that nanoparticles do not undergo burst release from devices post-implantation and that inflammatory responses - a key determinant of device function in vivo - are also unaffected by nanoparticle addition.


Subject(s)
Contrast Media , Inflammation , X-Ray Microtomography , Animals , Inflammation/pathology , Mice , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Polyesters/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Macrophages/metabolism , Nanoparticles/chemistry , Mice, Inbred C57BL
2.
Curr Opin Chem Biol ; 69: 102152, 2022 08.
Article in English | MEDLINE | ID: mdl-35561425

ABSTRACT

Faster, more sensitive, and higher resolution quantitative instrumentation are aiding a deeper understanding of how inorganic chemistry regulates key biological processes. Researchers can now image and quantify metals with subcellular resolution, leading to a vast array of new discoveries in organismal development, pathology, and disease. Metals have recently been implicated in several diseases such as Parkinson's, Alzheimers, ischemic stroke, and colorectal cancer that would not be possible without these advancements. In this review, instead of focusing on instrumentation we focus on recent applications of label-free elemental imaging and quantification and how these tools can lead to a broader understanding of metals role in systems biology and human pathology.


Subject(s)
Diagnostic Imaging , Metals , Diagnostic Imaging/methods , Humans , Ions , Mass Spectrometry/methods
3.
Nat Med ; 26(9): 1422-1427, 2020 09.
Article in English | MEDLINE | ID: mdl-32651581

ABSTRACT

Antibodies are a principal determinant of immunity for most RNA viruses and have promise to reduce infection or disease during major epidemics. The novel coronavirus SARS-CoV-2 has caused a global pandemic with millions of infections and hundreds of thousands of deaths to date1,2. In response, we used a rapid antibody discovery platform to isolate hundreds of human monoclonal antibodies (mAbs) against the SARS-CoV-2 spike (S) protein. We stratify these mAbs into five major classes on the basis of their reactivity to subdomains of S protein as well as their cross-reactivity to SARS-CoV. Many of these mAbs inhibit infection of authentic SARS-CoV-2 virus, with most neutralizing mAbs recognizing the receptor-binding domain (RBD) of S. This work defines sites of vulnerability on SARS-CoV-2 S and demonstrates the speed and robustness of advanced antibody discovery platforms.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/virology , Humans , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/immunology
4.
bioRxiv ; 2020 May 13.
Article in English | MEDLINE | ID: mdl-32511414

ABSTRACT

Antibodies are a principal determinant of immunity for most RNA viruses and have promise to reduce infection or disease during major epidemics. The novel coronavirus SARS-CoV-2 has caused a global pandemic with millions of infections and hundreds of thousands of deaths to date 1,2 . In response, we used a rapid antibody discovery platform to isolate hundreds of human monoclonal antibodies (mAbs) against the SARS-CoV-2 spike (S) protein. We stratify these mAbs into five major classes based on their reactivity to subdomains of S protein as well as their cross-reactivity to SARS-CoV. Many of these mAbs inhibit infection of authentic SARS-CoV-2 virus, with most neutralizing mAbs recognizing the receptor-binding domain (RBD) of S. This work defines sites of vulnerability on SARS-CoV-2 S and demonstrates the speed and robustness of new antibody discovery methodologies.

5.
Bioconjug Chem ; 30(11): 2947-2957, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31589412

ABSTRACT

ProGlo is an efficient steroid receptor-targeted magnetic resonance (MR) imaging contrast agent (CA). It has been shown to bind to the progesterone receptor (PR) and produce enhanced image contrast in PR-positive cells and tissues in vitro and in vivo. However, the hydrophobicity of the steroid targeting domain of ProGlo (logP = 1.4) limits its formulation and delivery at clinically relevant doses. In this work, a hydrophobic moiety was utilized to drive efficient adsorption onto nanodiamond (ND) clusters to form a water-soluble nanoconstruct (logP = -2.4) with 80% release in 8 h under biological conditions. In cell culture, the ND-ProGlo construct delivered increased concentrations of ProGlo to target cells compared to ProGlo alone. Importantly, these results were accomplished without the use of solvents such as DMSO, providing a significant advance toward formulating ProGlo for translational applications. Biodistribution studies confirm the delivery of ProGlo to PR(+) tissues with enhanced efficacy over untargeted controls. These results demonstrate the potential for a noncovalent ND-CA construct as a general strategy for solubilizing and delivering hydrophobic targeted MR CAs.


Subject(s)
Breast Neoplasms/pathology , Contrast Media/pharmacokinetics , Magnetic Resonance Imaging/methods , Nanoconjugates/chemistry , Nanodiamonds/administration & dosage , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/metabolism , Contrast Media/chemistry , Female , Humans , Mice , Nanodiamonds/chemistry , Receptors, Progesterone/chemistry , Solubility , Tissue Distribution , Tumor Cells, Cultured
6.
Bioconjug Chem ; 29(11): 3544-3549, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30193061

ABSTRACT

We describe a new, and vastly superior approach for labeling spherical nucleic acid conjugates (SNAs) with diagnostic probes. SNAs have been shown to provide the unique ability to traverse the cell membrane and deliver surface conjugated DNA into cells while preserving the DNA from nuclease degradation. Our previous work on preparing diagnostically labeled SNAs was labor intensive, relatively low yielding, and costly. Here, we describe a straightforward and facile preparation for labeling SNAs with optical and MR imaging probes with significantly improved physical properties. The synthesis of Gd(III) labeled DNA Au nanoparticle conjugates is achieved by sequential conjugation of 3'-thiol-modified oligonucleotides and cofunctionalization of the particle surface with the subsequent addition of 1,2 diothiolate modified chelates of Gd(III) (abbreviated: DNA-GdIII@AuNP). This new generation of SNA conjugates has a 2-fold increase of DNA labeling and a 1.4-fold increase in Gd(III) loading compared to published constructs. Furthermore, the relaxivity ( r1) is observed to increase 4.5-fold compared to the molecular dithiolane-Gd(III) complex, and 1.4-fold increase relative to previous particle constructs where the Gd(III) complexes were conjugated to the oligonucleotides rather than directly to the Au particle. Importantly, this simplified approach (2 steps) exploits the advantages of previous Gd(III) labeled SNA platforms; however, this new approach is scalable and eliminates modification of DNA for attaching the contrast agent, and the particles exhibit improved cell labeling.


Subject(s)
DNA/chemistry , Gold/chemistry , Magnetic Resonance Imaging/methods , Metal Nanoparticles/chemistry , Molecular Probes/chemistry , Contrast Media , Gadolinium/chemistry , Sulfhydryl Compounds/chemistry , Surface Properties
7.
Chem Sci ; 8(8): 5764-5768, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28989616

ABSTRACT

Visualizing disease heterogeneity remains a challenging task since most imaging agents are targeted to a single receptor. We describe the development of an MR platform able to report on multiple molecular events. Enzyme activation and enhanced cellular uptake of this modular probe make it suitable for subsequent targeted-reporter imaging applications.

8.
Sci Rep ; 7(1): 473, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28352089

ABSTRACT

A widely prevalent single nucleotide polymorphism, rs13266634 in the SLC30A8 gene encoding the zinc transporter ZnT8, is associated with an increased risk for T2DM. ZnT8 is mostly expressed in pancreatic insulin-producing islets of Langerhans. The effect of this variant on the divalent metal profile in human islets is unknown. Additionally, essential and non-essential divalent metal content of human islets under normal environmental exposure conditions has not been described. We therefore examined the correlation of zinc and other divalent metals in human islets with rs13266634 genotype and demographic characteristics. We found that the diabetes risk genotype C/C at rs13266634 is associated with higher islet Zn concentration (C/C genotype: 16792 ± 1607, n = 22, C/T genotype: 11221 ± 1245, n = 18 T/T genotype: 11543 ± 6054, n = 3, all values expressed as mean nmol/g protein ± standard error of the mean, p = 0.040 by ANOVA). A positive correlation between islet cadmium content and both age (p = 0.048, R2 = 0.09) and female gender (women: 36.88 ± 4.11 vs men: 21.22 ± 3.65 nmol/g protein, p = 0.007) was observed. Our results suggest that the T2DM risk allele C is associated with higher islet zinc levels and support prior evidence of cadmium's higher bioavailability in women and its long tissue half-life.


Subject(s)
Genotype , Islets of Langerhans/metabolism , Metals/metabolism , Polymorphism, Single Nucleotide , Zinc Transporter 8/genetics , Zinc/metabolism , Adult , Age Factors , Alleles , Cadmium/metabolism , Copper/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Iron/metabolism , Male , Manganese/metabolism , Middle Aged , Nickel/metabolism , Zinc Transporter 8/metabolism
9.
Bioconjug Chem ; 28(1): 153-160, 2017 01 18.
Article in English | MEDLINE | ID: mdl-27537821

ABSTRACT

In vivo cell tracking is vital for understanding migrating cell populations, particularly cancer and immune cells. Magnetic resonance (MR) imaging for long-term tracking of transplanted cells in live organisms requires cells to effectively internalize Gd(III) contrast agents (CAs). Clinical Gd(III)-based CAs require high dosing concentrations and extended incubation times for cellular internalization. To combat this, we have devised a series of Gd(III)-gold nanoconjugates (Gd@AuNPs) with varied chelate structure and nanoparticle-chelate linker length, with the goal of labeling and imaging breast cancer cells. These new Gd@AuNPs demonstrate significantly enhanced labeling compared to previous Gd(III)-gold-DNA nanoconstructs. Variations in Gd(III) loading, surface packing, and cell uptake were observed among four different Gd@AuNP formulations suggesting that linker length and surface charge play an important role in cell labeling. The best performing Gd@AuNPs afforded 23.6 ± 3.6 fmol of Gd(III) per cell at an incubation concentration of 27.5 µM-this efficiency of Gd(III) payload delivery (Gd(III)/cell normalized to dose) exceeds that of previous Gd(III)-Au conjugates and most other Gd(III)-nanoparticle formulations. Further, Gd@AuNPs were well-tolerated in vivo in terms of biodistribution and clearance, and supports future cell tracking applications in whole-animal models.


Subject(s)
Gadolinium/chemistry , Gold/chemistry , Magnetic Resonance Imaging/methods , Nanoconjugates/chemistry , Animals , Cell Line, Tumor , Humans , Mass Spectrometry , Mice
10.
Nano Lett ; 16(12): 7551-7564, 2016 12 14.
Article in English | MEDLINE | ID: mdl-27960515

ABSTRACT

The ability to track labeled cancer cells in vivo would allow researchers to study their distribution, growth, and metastatic potential within the intact organism. Magnetic resonance (MR) imaging is invaluable for tracking cancer cells in vivo as it benefits from high spatial resolution and the absence of ionizing radiation. However, many MR contrast agents (CAs) required to label cells either do not significantly accumulate in cells or are not biologically compatible for translational studies. We have developed carbon-based nanodiamond-gadolinium(III) aggregates (NDG) for MR imaging that demonstrated remarkable properties for cell tracking in vivo. First, NDG had high relaxivity independent of field strength, a finding unprecedented for gadolinium(III) [Gd(III)]-nanoparticle conjugates. Second, NDG demonstrated a 300-fold increase in the cellular delivery of Gd(III) compared to that of clinical Gd(III) chelates without sacrificing biocompatibility. Further, we were able to monitor the tumor growth of NDG-labeled flank tumors by T1- and T2-weighted MR imaging for 26 days in vivo, longer than was reported for other MR CAs or nuclear agents. Finally, by utilizing quantitative maps of relaxation times, we were able to describe tumor morphology and heterogeneity (corroborated by histological analysis), which would not be possible with competing molecular imaging modalities.


Subject(s)
Gadolinium , Molecular Imaging , Nanodiamonds , Neoplasms, Experimental/diagnostic imaging , Animals , Contrast Media , Female , Magnetic Resonance Imaging , Mice , Mice, SCID
11.
Nano Lett ; 16(5): 3202-9, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27050622

ABSTRACT

Pancreatic adenocarcinoma has a 5 year survival of approximately 3% and median survival of 6 months and is among the most dismal of prognoses in all of medicine. This poor prognosis is largely due to delayed diagnosis where patients remain asymptomatic until advanced disease is present. Therefore, techniques to allow early detection of pancreatic adenocarcinoma are desperately needed. Imaging of pancreatic tissue is notoriously difficult, and the development of new imaging techniques would impact our understanding of organ physiology and pathology with applications in disease diagnosis, staging, and longitudinal response to therapy in vivo. Magnetic resonance imaging (MRI) provides numerous advantages for these types of investigations; however, it is unable to delineate the pancreas due to low inherent contrast within this tissue type. To overcome this limitation, we have prepared a new Gd(III) contrast agent that accumulates in the pancreas and provides significant contrast enhancement by MR imaging. We describe the synthesis and characterization of a new dithiolane-Gd(III) complex and a straightforward and scalable approach for conjugation to a gold nanoparticle. We present data that show the nanoconjugates exhibit very high per particle values of r1 relaxivity at both low and high magnetic field strengths due to the high Gd(III) payload. We provide evidence of pancreatic tissue labeling that includes MR images, post-mortem biodistribution analysis, and pancreatic tissue evaluation of particle localization. Significant contrast enhancement was observed allowing clear identification of the pancreas with contrast-to-noise ratios exceeding 35:1.


Subject(s)
Contrast Media/chemistry , Gadolinium/chemistry , Gold/chemistry , Magnetic Resonance Imaging/methods , Metal Nanoparticles/chemistry , Pancreatic Neoplasms/diagnostic imaging , Sulfhydryl Compounds/chemistry , Animals , Gadolinium/pharmacokinetics , Heterografts , Humans , Male , Mice, Inbred C57BL , Nanoconjugates/chemistry , Particle Size , Surface Properties , Tissue Distribution , Pancreatic Neoplasms
12.
Biomaterials ; 77: 291-306, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26615367

ABSTRACT

The unambiguous imaging of transplanted cells remains a major challenge to understand their biological function and therapeutic efficacy. In vivo imaging of implanted cells is reliant on tagging these to differentiate them from host tissue, such as the brain. We here characterize a gold nanoparticle conjugate that is functionalized with modified deoxythymidine oligonucleotides bearing Gd(III) chelates and a red fluorescent Cy3 moiety to visualize in vivo transplanted human neural stem cells. This DNA-Gd@Au nanoparticle (DNA-Gd@AuNP) exhibits an improved T1 relaxivity and excellent cell uptake. No significant effects of cell uptake have been found on essential cell functions. Although T1 relaxivity is attenuated within cells, it is sufficiently preserved to afford the in vivo detection of transplanted cells using an optimized voxel size. In vivo MR images were corroborated by a post-mortem histological verification of DNA-Gd@AuNPs in transplanted cells. With 70% of cells being correctly identified using the DNA-Gd-AuNPs indicates an overall reliable detection. Less than 1% of cells were false positive for DNA-Gd@AuNPs, but a significant number of 30% false negatives reveals a dramatic underestimation of transplanted cells using this approach. DNA-Gd@AuNPs therefore offer new opportunities to visualize transplanted cells unequivocally using T1 contrast and use cellular MRI as a tool to derive biologically relevant information that allows us to understand how the survival and location of implanted cells determines therapeutic efficacy.


Subject(s)
Cell Tracking/methods , Contrast Media/analysis , DNA/analysis , Gadolinium/analysis , Gold Colloid/analysis , Magnetic Resonance Imaging/methods , Nanoconjugates/analysis , Nanoparticles/analysis , Neural Stem Cells/transplantation , Animals , Astrocytes/cytology , Cell Line , Cerebral Cortex/ultrastructure , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Corpus Striatum/cytology , DNA/administration & dosage , DNA/pharmacokinetics , Gadolinium/administration & dosage , Gadolinium/pharmacokinetics , Gold Colloid/administration & dosage , Gold Colloid/pharmacokinetics , Graft Survival , Humans , Neurogenesis , Neurons/cytology , Oligonucleotides/administration & dosage , Oligonucleotides/analysis , Oligonucleotides/chemical synthesis , Oligonucleotides/pharmacokinetics , Phantoms, Imaging , Signal-To-Noise Ratio , Thymidine
13.
Bioconjug Chem ; 27(2): 465-73, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26689452

ABSTRACT

Calcium [Ca(II)] is a fundamental transducer of electrical activity in the central nervous system (CNS). Influx of Ca(II) into the cytosol is responsible for action potential initiation and propagation, and initiates interneuronal communication via release of neurotransmitters and activation of gene expression. Despite the importance of Ca(II) in physiology, it remains a challenge to visualize Ca(II) flux in the central nervous system (CNS) in vivo. To address these challenges, we have developed a new generation, Ca(II)-activated MRI contrast agent that utilizes ethyl esters to increase cell labeling and prevent extracellular divalent Ca(II) binding. Following labeling, the ethyl esters can be cleaved, thus allowing the agent to bind Ca(II), increasing relaxivity and resulting in enhanced positive MR image contrast. The ability of this probe to discriminate between extra- and intracellular Ca(II) may allow for spatiotemporal in vivo imaging of Ca(II) flux during seizures or ischemia where large Ca(II) fluxes (1-10 µM) can result in cell death.


Subject(s)
Calcium/analysis , Contrast Media/metabolism , Esterases/metabolism , Magnetic Resonance Imaging , Animals , Calcium/metabolism , Cell Line , Cell Membrane Permeability , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Extracellular Space/chemistry , Extracellular Space/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Intracellular Space/chemistry , Intracellular Space/metabolism , Magnetic Resonance Imaging/methods , Mice
15.
J Biol Inorg Chem ; 20(6): 971-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26215869

ABSTRACT

Long-term cell tracking using MR imaging necessitates the development of contrast agents that both label and are retained by cells. One promising strategy for long-term cell labeling is the development of lipophilic Gd(III)-based contrast agents that anchor into the cell membrane. We have previously reported the efficacy of monomeric and multimeric lipophilic agents and showed that the monomeric agents have improved labeling and contrast enhancement of cell populations. Here, we report on the synthesis, characterization, and in vitro testing of a series of monomeric lipophilic contrast agents with varied alkyl chain compositions. We show that these agents disperse in water, localize to the cell membrane, and label HeLa and MCF7 cells effectively. Additionally, these agents have up to tenfold improved retention in cells compared to clinically available ProHance(®).


Subject(s)
Cell Membrane/metabolism , Contrast Media/chemical synthesis , Coordination Complexes/chemical synthesis , Gadolinium/chemistry , Cell Membrane/chemistry , Contrast Media/toxicity , Coordination Complexes/toxicity , Gadolinium/toxicity , HeLa Cells , Humans , MCF-7 Cells , Magnetic Resonance Imaging
16.
J Am Chem Soc ; 137(28): 9108-16, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26083313

ABSTRACT

Multiple imaging modalities are often required for in vivo imaging applications that require both high probe sensitivity and excellent spatial and temporal resolution. In particular, MR and optical imaging are an attractive combination that can be used to determine both molecular and anatomical information. Herein, we describe the synthesis and in vivo testing of two multimeric NIR-MR contrast agents that contain three Gd(III) chelates and an IR-783 dye moiety. One agent contains a PEG linker and the other a short alkyl linker. These agents label cells with extraordinary efficacy and can be detected in vivo using both imaging modalities. Biodistribution of the PEGylated agent shows observable fluorescence in xenograft MCF7 tumors and renal clearance by MR imaging.


Subject(s)
Chelating Agents/chemistry , Coloring Agents/chemistry , Contrast Media/chemistry , Gadolinium/chemistry , Magnetic Resonance Imaging , Multimodal Imaging , Optical Imaging , Animals , Chelating Agents/chemical synthesis , Chelating Agents/pharmacokinetics , Coloring Agents/chemical synthesis , Coloring Agents/pharmacokinetics , Contrast Media/chemical synthesis , Contrast Media/pharmacokinetics , Female , Gadolinium/pharmacokinetics , Humans , Infrared Rays , MCF-7 Cells , Mice, Nude , Neoplasms/diagnosis , Tissue Distribution
17.
Bioconjug Chem ; 26(5): 899-905, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25830565

ABSTRACT

Nanodiscs are monodisperse, self-assembled discoidal particles that consist of a lipid bilayer encircled by membrane scaffold proteins (MSP). Nanodiscs have been used to solubilize membrane proteins for structural and functional studies and deliver therapeutic phospholipids. Herein, we report on tetramethylrhodamine (TMR) tagged nanodiscs that solubilize lipophilic MR contrast agents for generation of multimodal nanoparticles for cellular imaging. We incorporate both multimeric and monomeric Gd(III)-based contrast agents into nanodiscs and show that particles containing the monomeric agent (ND2) label cells with high efficiency and generate significant image contrast at 7 T compared to nanodiscs containing the multimeric agent (ND1) and Prohance, a clinically approved contrast agent.


Subject(s)
Magnetic Resonance Imaging/methods , Multimodal Imaging/methods , Nanostructures/chemistry , Optical Imaging/methods , Contrast Media/chemistry , Gadolinium/chemistry , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Models, Molecular , Molecular Conformation , Rhodamines/chemistry , Solubility
18.
ACS Nano ; 9(3): 3385-96, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25723190

ABSTRACT

Gadolinium(III) nanoconjugate contrast agents (CAs) have distinct advantages over their small-molecule counterparts in magnetic resonance imaging. In addition to increased Gd(III) payload, a significant improvement in proton relaxation efficiency, or relaxivity (r1), is often observed. In this work, we describe the synthesis and characterization of a nanoconjugate CA created by covalent attachment of Gd(III) to thiolated DNA (Gd(III)-DNA), followed by surface conjugation onto gold nanostars (DNA-Gd@stars). These conjugates exhibit remarkable r1 with values up to 98 mM(-1) s(-1). Additionally, DNA-Gd@stars show efficient Gd(III) delivery and biocompatibility in vitro and generate significant contrast enhancement when imaged at 7 T. Using nuclear magnetic relaxation dispersion analysis, we attribute the high performance of the DNA-Gd@stars to an increased contribution of second-sphere relaxivity compared to that of spherical CA equivalents (DNA-Gd@spheres). Importantly, the surface of the gold nanostar contains Gd(III)-DNA in regions of positive, negative, and neutral curvature. We hypothesize that the proton relaxation enhancement observed results from the presence of a unique hydrophilic environment produced by Gd(III)-DNA in these regions, which allows second-sphere water molecules to remain adjacent to Gd(III) ions for up to 10 times longer than diffusion. These results establish that particle shape and second-sphere relaxivity are important considerations in the design of Gd(III) nanoconjugate CAs.


Subject(s)
Contrast Media/chemistry , DNA/chemistry , Gadolinium/chemistry , Gold/chemistry , Nanostructures/chemistry , Protons , Biological Transport , Cell Line, Tumor , Contrast Media/metabolism , Contrast Media/pharmacology , Humans , Magnetic Resonance Imaging , Materials Testing , Models, Molecular , Nucleic Acid Conformation
19.
Science ; 347(6223): 746-50, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25678658

ABSTRACT

Dental enamel, a hierarchical material composed primarily of hydroxylapatite nanowires, is susceptible to degradation by plaque biofilm-derived acids. The solubility of enamel strongly depends on the presence of Mg(2+), F(-), and CO3(2-). However, determining the distribution of these minor ions is challenging. We show­using atom probe tomography, x-ray absorption spectroscopy, and correlative techniques­that in unpigmented rodent enamel, Mg(2+) is predominantly present at grain boundaries as an intergranular phase of Mg-substituted amorphous calcium phosphate (Mg-ACP). In the pigmented enamel, a mixture of ferrihydrite and amorphous iron-calcium phosphate replaces the more soluble Mg-ACP, rendering it both harder and more resistant to acid attack. These results demonstrate the presence of enduring amorphous phases with a dramatic influence on the physical and chemical properties of the mature mineralized tissue.


Subject(s)
Calcium Phosphates/chemistry , Dental Enamel/chemistry , Dental Enamel/ultrastructure , Animals , Incisor/chemistry , Incisor/ultrastructure , Mice , Microscopy, Electron, Scanning , X-Ray Absorption Spectroscopy
20.
ACS Nano ; 8(10): 10168-77, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25226566

ABSTRACT

The delivery of bioactive molecules into cells has broad applications in biology and medicine. Polymer-modified graphene oxide (GO) has recently emerged as a de facto noncovalent vehicle for hydrophobic drugs. Here, we investigate a different approach using native GO to deliver hydrophilic molecules by co-incubation in culture. GO adsorption and delivery were systematically studied with a library of 15 molecules synthesized with Gd(III) labels to enable quantitation. Amines were revealed to be a key chemical group for adsorption, while delivery was shown to be quantitatively predictable by molecular adsorption, GO sedimentation, and GO size. GO co-incubation was shown to enhance delivery by up to 13-fold and allowed for a 100-fold increase in molecular incubation concentration compared to the alternative of nanoconjugation. When tested in the application of Gd(III) cellular MRI, these advantages led to a nearly 10-fold improvement in sensitivity over the state-of-the-art. GO co-incubation is an effective method of cellular delivery that is easily adoptable by researchers across all fields.


Subject(s)
Graphite/chemistry , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Imaging , Oxides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...